Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.528
Filter
1.
J Antimicrob Chemother ; 79(5): 1169-1175, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38546795

ABSTRACT

BACKGROUND: Invasive aspergillosis is a severe fungal infection that affects multiple organ systems including the CNS and the lungs. Isavuconazole, a novel triazole antifungal agent, has demonstrated promising activity against Aspergillus spp. However, data on the penetration of isavuconazole into the CNS and ELF and intracellular accumulation remain limited. MATERIALS AND METHODS: We conducted a prospective single-centre pharmacokinetic (PK) study in 12 healthy volunteers. Subjects received seven doses of 200 mg isavuconazole to achieve an assumed steady-state. After the first and final infusion, plasma sampling was conducted over 8 and 12 h, respectively. All subjects underwent one lumbar puncture and bronchoalveolar lavage, at either 2, 6 or 12 h post-infusion of the final dose. PBMCs were collected in six subjects from blood to determine intracellular isavuconazole concentrations at 6, 8 or 12 h. The AUC/MIC was calculated for an MIC value of 1 mg/L, which marks the EUCAST susceptibility breakpoint for Aspergillus fumigatus and Aspergillus flavus. RESULTS: C max and AUC0-24h of isavuconazole in plasma under assumed steady-state conditions were 6.57 ±â€Š1.68 mg/L (mean ±â€ŠSD) and 106 ±â€Š32.1 h·mg/L, respectively. The average concentrations measured in CSF, ELF and in PBMCs were 0.07 ±â€Š0.03, 0.94 ±â€Š0.46 and 27.1 ±â€Š17.8 mg/L, respectively. The AUC/MIC in plasma, CSF, ELF and in PBMCs under steady-state conditions were 106 ±â€Š32.1, 1.68 ±â€Š0.72, 22.6 ±â€Š11.0 and 650 ±â€Š426 mg·h/L, respectively. CONCLUSION: Isavuconazole demonstrated moderate penetration into ELF, low penetrability into CSF and high accumulation in PBMCs. Current dosing regimens resulted in sufficient plasma exposure in all subjects to treat isolates with MICs ≤ 1 mg/L.


Subject(s)
Antifungal Agents , Healthy Volunteers , Nitriles , Pyridines , Triazoles , Humans , Triazoles/pharmacokinetics , Triazoles/administration & dosage , Pyridines/pharmacokinetics , Pyridines/administration & dosage , Antifungal Agents/pharmacokinetics , Antifungal Agents/administration & dosage , Male , Adult , Nitriles/pharmacokinetics , Nitriles/administration & dosage , Prospective Studies , Female , Infusions, Intravenous , Young Adult , Microbial Sensitivity Tests , Middle Aged , Aspergillus fumigatus/drug effects , Aspergillus flavus/drug effects , Bronchoalveolar Lavage Fluid/chemistry , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects
2.
Vet Rec ; 194(9): e3955, 2024 05 04.
Article in English | MEDLINE | ID: mdl-38462781

ABSTRACT

BACKGROUND: A single-dose, in-clinic, veterinary professional-administered treatment for canine otitis externa was developed to improve compliance and canine welfare. METHODS: This multicentre, controlled, examiner-masked, randomised field trial was conducted in 316 dogs over 42 days. Dogs were treated once, on day 0, with the investigational product containing gentamicin, posaconazole and mometasone furoate (Mometamax Ultra [MU]) or twice (days 0 and 7) with a control product containing florfenicol, terbinafine and betamethasone acetate (CP). The primary endpoint was a composite otitis index score of 4 or less (of 12) on day 14 and 3 or less (of 12) on day 28. RESULTS: On day 28, treatment success was recorded in 128 of 143 MU-treated dogs (89.5%), significantly non-inferior to 116 of 133 (87.2%) CP-treated dogs (Farrington-Manning test, Z = 4.1351, p < 0.0001). For mixed cultures of Staphylococcus pseudintermedius and Malassezia pachydermatis, there was 100% treatment success in MU-treated dogs (n = 33), significantly non-inferior to 90.2% (37 of 41) in CP-treated dogs (Farrington-Manning test, Z = 3.1954, p = 0.0007). LIMITATIONS: Efficacy in chronic otitis externa cases was not investigated. Cytology was not used to aid in diagnosis or for identification of secondary pathogens. CONCLUSION: This unique combination, single-dose product is safe and effective in dogs with otitis externa. It offers enhanced compliance, canine welfare and quality of life by eliminating the owner burden of treating this painful condition.


Subject(s)
Dog Diseases , Gentamicins , Mometasone Furoate , Otitis Externa , Triazoles , Animals , Dogs , Dog Diseases/drug therapy , Otitis Externa/veterinary , Otitis Externa/drug therapy , Otitis Externa/microbiology , Mometasone Furoate/therapeutic use , Mometasone Furoate/administration & dosage , Treatment Outcome , Female , Male , Triazoles/therapeutic use , Triazoles/administration & dosage , Gentamicins/therapeutic use , Gentamicins/administration & dosage , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/administration & dosage , Drug Therapy, Combination/veterinary , Antifungal Agents/therapeutic use , Antifungal Agents/administration & dosage , Suspensions
3.
Biomed Pharmacother ; 174: 116492, 2024 May.
Article in English | MEDLINE | ID: mdl-38537579

ABSTRACT

Targeting epigenetic mechanisms has emerged as a potential therapeutic approach for the treatment of kidney diseases. Specifically, inhibiting the bromodomain and extra-terminal (BET) domain proteins using the small molecule inhibitor JQ1 has shown promise in preclinical models of acute kidney injury (AKI) and chronic kidney disease (CKD). However, its clinical translation faces challenges due to issues with poor pharmacokinetics and side effects. Here, we developed engineered liposomes loaded with JQ1 with the aim of enhancing kidney drug delivery and reducing the required minimum effective dose by leveraging cargo protection. These liposomes efficiently encapsulated JQ1 in both the membrane and core, demonstrating superior therapeutic efficacy compared to freely delivered JQ1 in a mouse model of kidney ischemia-reperfusion injury. JQ1-loaded liposomes (JQ1-NPs) effectively targeted the kidneys and only one administration, one-hour after injury, was enough to decrease the immune cell (neutrophils and monocytes) infiltration to the kidney-an early and pivotal step to prevent damage progression. By inhibiting BRD4, JQ1-NPs suppress the transcription of pro-inflammatory genes, such as cytokines (il-6) and chemokines (ccl2, ccl5). This success not only improved early the kidney function, as evidenced by decreased serum levels of BUN and creatinine in JQ1-NPs-treated mice, along with reduced tissue expression of the damage marker, NGAL, but also halted the production of extracellular matrix proteins (Fsp-1, Fn-1, α-SMA and Col1a1) and the fibrosis development. In summary, this work presents a promising nanotherapeutic strategy for AKI treatment and its progression and provides new insights into renal drug delivery.


Subject(s)
Azepines , Bromodomain Containing Proteins , Disease Progression , Kidney , Liposomes , Mice, Inbred C57BL , Nuclear Proteins , Renal Insufficiency, Chronic , Reperfusion Injury , Triazoles , Animals , Azepines/pharmacology , Azepines/administration & dosage , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Triazoles/pharmacology , Triazoles/administration & dosage , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Mice , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Male , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Disease Models, Animal , Nanoparticles , Cell Cycle Proteins/antagonists & inhibitors
4.
Drugs R D ; 24(1): 97-108, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38472696

ABSTRACT

BACKGROUND AND OBJECTIVES: Daridorexant, a dual orexin receptor antagonist was recently approved for the treatment of insomnia at doses up to 50 mg once per night. This study investigated the effect of single-dose and multiple-dose daridorexant 50 mg at steady state on the pharmacokinetics (PK) of the cytochrome P450 (CYP) 3A4-sensitive substrate midazolam, and the effect of single-dose daridorexant 50 mg on the PK and pharmacodynamics (PD) of the CYP2C9-sensitive substrate warfarin. METHODS: In this prospective, single-center, open-label, fixed-sequence, phase I, drug-drug interaction study, 18 healthy male subjects sequentially received Treatment A, B, and C in three periods. Treatment A consisted of a single oral concomitant administration of midazolam 2 mg and warfarin 25 mg on day 1 of the first period. Treatment B consisted of one oral administration of daridorexant 50 mg followed 1 h later by a single oral dose of midazolam 2 mg concomitantly with a single oral dose of warfarin 25 mg on day 1 and a once-daily oral administration of daridorexant 50 mg for 6 days of the second period. Treatment C consisted of a single oral administration of daridorexant 50 mg at steady state followed 1 h later by a single oral administration of midazolam 2 mg on day 1 of the third period. Blood samples were assessed for midazolam and S-warfarin PK, and PD (international normalized ratio and factor VII). Noncompartmental  PK parameters and PD variables were evaluated with geometric mean ratios and 90% confidence intervals of Treatment B/A versus C/A for midazolam, and treatment B/A for warfarin. Safety and tolerability of each treatment were also assessed. RESULTS: Midazolam maximum plasma concentration (Cmax) and area under the plasma concentration-time curve from 0 to 24 h (AUC0-24) were 1.13- and 1.42-fold higher, respectively, after single-dose administration of daridorexant 50 mg compared to administration of midazolam alone, while Cmax and AUC0-24 were 1.12- and 1.35-fold higher, respectively, after administration of daridorexant 50 mg once daily at steady state. Terminal half-life and time to maximum plasma concentration were comparable between treatments. Daridorexant had no influence on the PK and PD of warfarin. All treatments were safe and well tolerated. CONCLUSIONS: Daridorexant at 50 mg is classified as a weak CYP3A4 inhibitor after single- and multiple-dose administration once daily at steady state. Daridorexant 50 mg did not induce CYP3A4 activity or inhibit CYP2C9 activity. CLINICAL TRIAL REGISTRATION: This trial (NCT05480488) was registered on 29 July, 2022.


Subject(s)
Drug Interactions , Imidazoles , Midazolam , Pyrrolidines , Warfarin , Humans , Male , Midazolam/pharmacokinetics , Midazolam/administration & dosage , Adult , Warfarin/pharmacokinetics , Warfarin/administration & dosage , Warfarin/pharmacology , Young Adult , Healthy Volunteers , Triazoles/pharmacokinetics , Triazoles/administration & dosage , Triazoles/pharmacology , Prospective Studies , Orexin Receptor Antagonists/pharmacokinetics , Orexin Receptor Antagonists/pharmacology , Orexin Receptor Antagonists/administration & dosage , Area Under Curve
5.
Clin Cancer Res ; 30(9): 1984-1991, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38376912

ABSTRACT

PURPOSE: BCI (H/I) has been shown to predict extended endocrine therapy (EET) benefit. We examined BCI (H/I) for EET benefit prediction in NSABP B-42, which evaluated extended letrozole therapy (ELT) in patients with hormone receptor-positive breast cancer after 5 years of ET. EXPERIMENTAL DESIGN: A stratified Cox model was used to analyze RFI as the primary endpoint, with DR, BCFI, and DFS as secondary endpoints. Because of a nonproportional effect of ELT on DR, time-dependent analyses were performed. RESULTS: The translational cohort included 2,178 patients (45% BCI (H/I)-High, 55% BCI (H/I)-Low). ELT showed an absolute 10-year RFI benefit of 1.6% (P = 0.10), resulting in an underpowered primary analysis (50% power). ELT benefit and BCI (H/I) did not show a significant interaction for RFI (BCI (H/I)-Low: 10 years absolute benefit 1.1% [HR, 0.70; 95% confidence interval (CI), 0.43-1.12; P = 0.13]; BCI (H/I)-High: 2.4% [HR, 0.83; 95% CI, 0.55-1.26; P = 0.38]; Pinteraction = 0.56). Time-dependent DR analysis showed that after 4 years, BCI (H/I)-High patients had significant ELT benefit (HR = 0.29; 95% CI, 0.12-0.69; P < 0.01), whereas BCI (H/I)-Low patients were less likely to benefit (HR, 0.68; 95% CI, 0.33-1.39; P = 0.29; Pinteraction = 0.14). Prediction of ELT benefit by BCI (H/I) was more apparent in the HER2- subset after 4 years (ELT-by-BCI (H/I) Pinteraction = 0.04). CONCLUSIONS: BCI (H/I)-High versus BCI (H/I)-Low did not show a statistically significant difference in ELT benefit for the primary endpoint (RFI). However, in time-dependent DR analysis, BCI (H/I)-High patients experienced statistically significant benefit from ELT after 4 years, whereas (H/I)-Low patients did not. Because BCI (H/I) has been validated as a predictive marker of EET benefit in other trials, additional follow-up may enable further characterization of BCI's predictive ability.


Subject(s)
Aromatase Inhibitors , Breast Neoplasms , Letrozole , Receptors, Estrogen , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Aromatase Inhibitors/therapeutic use , Middle Aged , Receptors, Estrogen/metabolism , Letrozole/therapeutic use , Letrozole/administration & dosage , Aged , Receptors, Progesterone/metabolism , Adult , Treatment Outcome , Nitriles/therapeutic use , Triazoles/therapeutic use , Triazoles/administration & dosage , Prognosis
6.
Clin Transl Med ; 13(12): e1513, 2023 12.
Article in English | MEDLINE | ID: mdl-38131168

ABSTRACT

BACKGROUND: The majority of pancreatic ductal adenocarcinoma (PDAC) patients experience disease progression while on treatment with gemcitabine and nanoparticle albumin-bound (nab)-paclitaxel (GemPac) necessitating the need for a more effective treatment strategy for this refractory disease. Previously, we have demonstrated that nuclear exporter protein exportin 1 (XPO1) is a valid therapeutic target in PDAC, and the selective inhibitor of nuclear export selinexor (Sel) synergistically enhances the efficacy of GemPac in pancreatic cancer cells, spheroids and patient-derived tumours, and had promising activity in a phase I study. METHODS: Here, we investigated the impact of selinexor-gemcitabine-nab-paclitaxel (Sel-GemPac) combination on LSL-KrasG12D/+ ; LSL-Trp53R172H/+ ; Pdx1-Cre (KPC) mouse model utilising digital spatial profiling (DSP) and single nuclear RNA sequencing (snRNAseq). RESULTS: Sel-GemPac synergistically inhibited the growth of the KPC tumour-derived cell line. The Sel-GemPac combination reduced the 2D colony formation and 3D spheroid formation. In the KPC mouse model, at a sub-maximum tolerated dose (sub-MTD) , Sel-GemPac enhanced the survival of treated mice compared to controls (p < .05). Immunohistochemical analysis of residual KPC tumours showed re-organisation of tumour stromal architecture, suppression of proliferation and nuclear retention of tumour suppressors, such as Forkhead Box O3a (FOXO3a). DSP revealed the downregulation of tumour promoting genes such as chitinase-like protein 3 (CHIL3/CHI3L3/YM1) and multiple pathways including phosphatidylinositol 3'-kinase-Akt (PI3K-AKT) signalling. The snRNAseq demonstrated a significant loss of cellular clusters in the Sel-GemPac-treated mice tumours including the CD44+ stem cell population. CONCLUSION: Taken together, these results demonstrate that the Sel-GemPac treatment caused broad perturbation of PDAC-supporting signalling networks in the KPC mouse model. HIGHLIGHTS: The majority of pancreatic ductal adenocarcinoma (PDAC) patients experience disease progression while on treatment with gemcitabine and nanoparticle albumin-bound (nab)-paclitaxel (GemPac). Exporter protein exportin 1 (XPO1) inhibitor selinexor (Sel) with GemPac synergistically inhibited the growth of LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1-Cre (KPC) mouse derived cell line and enhanced the survival of mice. Digital spatial profiling shows that Sel-GemPac causes broad perturbation of PDAC-supporting signalling in the KPC model.


Subject(s)
Carcinoma, Pancreatic Ductal , Drug Combinations , Exportin 1 Protein , Pancreatic Neoplasms , Animals , Mice , Disease Models, Animal , Pancreatic Neoplasms/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Exportin 1 Protein/antagonists & inhibitors , Gemcitabine/administration & dosage , Paclitaxel/administration & dosage , Hydrazines/administration & dosage , Triazoles/administration & dosage , Tumor Microenvironment , Single-Cell Gene Expression Analysis , Humans
7.
J Med Chem ; 65(24): 16665-16678, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36512715

ABSTRACT

In our continuing efforts to discover novel triazoles with improved antifungal activity in vitro and in vivo, a series of 41 novel compounds containing 1,2,3-triazole side chains were designed and synthesized via a click reaction based on our previous work. Most of the compounds showed moderate to excellent broad-spectrum antifungal activity in vitro. Among them, the most promising compound 9A16 displayed excellent antifungal and anti-drug-resistant fungal ability (MIC80 = 0.0156-8 µg/mL). In addition, compound 9A16 showed powerful in vivo efficacy on mice systematically infected with Candida albicans SC5314, Cryptococcus neoformans H99, fluconazole-resistant C. albicans 100, and Aspergillus fumigatus 7544. Moreover, compared to fluconazole, compound 9A16 showed better in vitro anti-biofilm activity and was more difficult to induce drug resistance in a 1 month induction of resistance assay in C. albicans. With favorable pharmacokinetics, an acceptable safety profile, and high potency in vitro and in vivo, compound 9A16 is currently under preclinical investigation.


Subject(s)
Antifungal Agents , Triazoles , Animals , Mice , Antifungal Agents/administration & dosage , Antifungal Agents/chemistry , Antifungal Agents/pharmacokinetics , Candida albicans/drug effects , Fluconazole/pharmacology , Microbial Sensitivity Tests , Triazoles/administration & dosage , Triazoles/chemistry , Triazoles/pharmacokinetics , Administration, Oral , Biological Availability
8.
Lancet Psychiatry ; 9(3): 199-210, 2022 03.
Article in English | MEDLINE | ID: mdl-35151410

ABSTRACT

BACKGROUND: There are no approved pharmacological therapies to support treatment of the core communication and socialisation difficulties associated with autism spectrum disorder in adults. We aimed to assess the efficacy, safety, and pharmacokinetics of balovaptan, a vasopressin 1a receptor antagonist, versus placebo in autistic adults. METHODS: V1aduct was a phase 3, randomised, placebo-controlled, double-blind trial, conducted at 46 sites across six countries (the USA, the UK, France, Italy, Spain, and Canada). Eligible participants were aged 18 years or older with an intelligence quotient (IQ) of 70 or higher, and met the criteria for moderate-to-severe autism spectrum disorder (DSM-5 and Autism Diagnostic Observation Schedule). Participants were randomly allocated (1:1), with an independent interactive voice or web-based response system, to receive balovaptan (10 mg) or placebo daily for 24 weeks. Randomisation was stratified by an individual's baseline Vineland-II two-domain composite (2DC) score (<60 or ≥60), sex, region (North America or rest of world), and age (<25 years or ≥25 years). Participants, study site personnel, and the sponsor were masked to treatment assignment. The primary endpoint was change from baseline in Vineland-II 2DC score (the mean composite score across the Vineland-II socialisation and communication domains) at week 24. The primary analysis was done with ANCOVA in the intention-to-treat population. The V1aduct study was terminated for futility after around 50% of participants completed the week 24 visit. This trial is registered with ClinicalTrials.gov (NCT03504917). FINDINGS: Between Aug 8, 2018, and July 1, 2020, 540 people were screened for eligibility, of whom 322 were allocated to receive balovaptan (164 [51%]) or placebo (158 [49%]). One participant from the balovaptan group was not treated before trial termination and was excluded from the analysis. 60 participants in the balovaptan group and 55 in the placebo group discontinued treatment before week 24. The sample consisted of 64 (20%) women and 257 (80%) men, with 260 (81%) participants from North America and 61 (19%) from Europe. At baseline, mean age was 27·6 years (SD 9·7) and mean IQ score was 104·8 (18·1). Two (1%) participants were American Indian or Alaska Native, eight (2%) were Asian, 15 (5%) were Black or African American, 283 (88%) were White, four (1%) were of multiple races, and nine (3%) were of unknown race. Mean baseline Vineland-II 2DC scores were 67·2 (SD 15·3) in the balovaptan group and 66·2 (17·7) in the placebo group. The interim futility analysis showed no improvement for balovaptan versus placebo in terms of Vineland-II 2DC score at week 24 compared with baseline, with a least-squares mean change of 2·91 (SE 1·52) in the balovaptan group (n=79) and 4·75 (1·60) in the placebo group (n=71; estimated treatment difference -1·84 [95% CI -5·15 to 1·48]). In the final analysis, mean change from baseline in Vineland-II 2DC score at week 24 was 4·56 (SD 10·85) in the balovaptan group (n=111) and 6·83 (12·18) in the placebo group (n=99). Balovaptan was well tolerated, with similar proportions of participants with at least one adverse event in the balovaptan group (98 [60%] of 163) and placebo group (104 [66%] of 158). The most common adverse events were nasopharyngitis (14 [9%] in the balovaptan group and 19 [12%] in the placebo group), diarrhoea (11 [7%] and 14 [9%]), upper respiratory tract infection (ten [6%] and nine [6%]), insomnia (five [3%] and eight [5%]), oropharyngeal pain (five [3%] and eight [5%]), and dizziness (two [1%] and ten [6%]). Serious adverse events were reported for two (1%) participants in the balovaptan group (one each of suicidal ideation and schizoaffective disorder), and five (3%) participants in the placebo group (one each of suicidal ideation, panic disorder, limb abscess, urosepsis, colitis [in the same participant with urosepsis], and death by suicide). No treatment-related deaths occurred. INTERPRETATION: Balovaptan did not improve social communication in autistic adults. This study provides insights into challenges facing autism spectrum disorder trials, including the considerable placebo response and the selection of appropriate outcome measures. FUNDING: F Hoffmann-La Roche.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/administration & dosage , Autism Spectrum Disorder/drug therapy , Benzodiazepines/administration & dosage , Communication Disorders/drug therapy , Pyridines/administration & dosage , Triazoles/administration & dosage , Adult , Antidiuretic Hormone Receptor Antagonists/adverse effects , Autism Spectrum Disorder/complications , Benzodiazepines/adverse effects , Communication Disorders/etiology , Double-Blind Method , Female , Humans , Male , Pyridines/adverse effects , Treatment Outcome , Triazoles/adverse effects
9.
Arch Toxicol ; 96(2): 559-570, 2022 02.
Article in English | MEDLINE | ID: mdl-35048155

ABSTRACT

Prothioconazole (PTC) is a new broad-spectrum triazole antibacterial agent that is being widely used in agriculture. PTC has been linked to a number of reproductive outcomes including embryo implantation disorder; however, the exact mechanism underlying this relationship has yet to be determined. Proper trophoblast proliferation and migration is a prerequisite for successful embryo implantation. To elucidate the underlying molecular perturbations, we detect the effect of PTC on extravillous trophoblast cells proliferation and migration, and investigate its potential mechanisms. Exposure to different concentrations of PTC (0-500 µM) significantly inhibited the cell viability and migration ability (5 µM PTC exposure), and also caused the cell cycle arrest at the lowest dose (1 µM PTC exposure). Transcriptome analysis revealed that PTC exposure disturbed multiple biological processes including cell cycle and apoptosis, consistent with cell phenotype. Specifically, eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1, 4E-BP1) was identified as up-regulated in PTC exposure group and knockdown of EIF4EBP1, and attenuated the G1 phase arrest induced by PTC exposure. In summary, our data demonstrated that 4E-BP1 participated in PTC-induced cell cycle arrest in extravillous trophoblast cells by regulating cyclin D1. These findings shed light on the potential adverse effect of PTC exposure on the embryo implantation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Triazoles/toxicity , Trophoblasts/drug effects , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Cyclin D1/metabolism , Dose-Response Relationship, Drug , Female , Fungicides, Industrial/administration & dosage , Fungicides, Industrial/toxicity , Gene Knockdown Techniques , Humans , Triazoles/administration & dosage , Trophoblasts/cytology , Up-Regulation/drug effects
10.
AAPS J ; 24(1): 21, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34988679

ABSTRACT

While the concept of 'Virtual Bioequivalence' (VBE) using a combination of modelling, in vitro tests and integration of pre-existing data on systems and drugs is growing from its infancy, building confidence on VBE outcomes requires demonstration of its ability not only in predicting formulation-dependent systemic exposure but also the expected degree of population variability. The concept of variation influencing the outcome of BE, despite being hidden with the cross-over nature of common BE studies, becomes evident when dealing with the acceptance criteria that consider the 90% confidence interval (CI) around the relative bioavailability. Hence, clinical studies comparing a reference product against itself may fail due to within-subject variations associated with the two occasions that the individual receives the same formulation. In this proof-of-concept study, we offer strategies to capture the most realistic predictions of CI around the pharmacokinetic parameters by propagating physiological variations through physiologically based pharmacokinetic modelling. The exercise indicates feasibility of the approach based on comparisons made between the simulated and observed WSV of pharmacokinetic parameters tested for a clinical bioequivalence case study. However, it also indicates that capturing WSV of a large array of physiological parameters using backward translation modelling from repeated BE studies of reference products would require a diverse set of drugs and formulations. The current case study of delayed-release formulation of posaconazole was able to declare certain combinations of WSV of physiological parameters as 'not plausible'. The eliminated sets of WSV values would be applicable to PBPK models of other drugs and formulations. Graphical Abstract.


Subject(s)
Models, Biological , Research Design , Triazoles/administration & dosage , Adolescent , Adult , Biological Availability , Cross-Over Studies , Delayed-Action Preparations , Gastrointestinal Tract/physiology , Humans , Male , Middle Aged , Proof of Concept Study , Therapeutic Equivalency , Triazoles/pharmacokinetics , Young Adult
11.
Eur J Pharm Biopharm ; 171: 39-49, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34998911

ABSTRACT

Bromodomain and extraterminal domain protein inhibitors (BETi) for cancer treatment did not convince during their first clinical trials. Their epigenetic mechanism of action is still not well understood, even if MYC is generally considered as its main downstream target. In this context, we intended to assess two new nanoformulations of the BETi JQ1 for the treatment of colorectal cancer (CRC). JQ1 was encapsulated at 10 mg/mL in lipid nanocapsules (LNC) or polymeric micelles (PM), both compatible for an intravenous administration. Their effect was compared with free JQ1 on several CRC cell lines in vitro and with daily intraperitoneal cyclodextrin (CD)-loaded JQ1 on the CT26 CRC tumor model in vivo. We showed that LNC preferentially accumulated in tumor, liver, and lymph nodes. LNC-JQ1 and CD-JQ1 similarly delayed tumor growth and increased median survival from 15 to 23 or 20.5 days. JQ1 altered MYC in only two among four CRC cell lines. This MYC-independence found in CT26 was confirmed in vivo by PCR and immunohistochemistry. The main explanation of the JQ1 anticancer effect was an increase in apoptosis. The investigation of its impact on the tumor microenvironment did not show significant effects. Finally, JQ1 association with irinotecan did not synergize in vivo with JQ1 nanoformulations. In conclusion, we demonstrated that the JQ1 anticancer effect was not improved by nanoencapsulation even if their tumor delivery was probably higher. MYC inhibition was not associated to JQ1 efficacy in the case of the CT26 CRC murine model.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , Colorectal Neoplasms/drug therapy , Liposomes , Nanoparticles , Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Azepines/administration & dosage , Azepines/therapeutic use , Cell Line, Tumor/drug effects , Colorectal Neoplasms/metabolism , Drug Delivery Systems , Female , Humans , Infusions, Intravenous , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-myc/metabolism , Triazoles/administration & dosage , Triazoles/therapeutic use
12.
Clin Cancer Res ; 28(3): 452-460, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34728525

ABSTRACT

PURPOSE: Selinexor is an oral selective inhibitor of exportin-1 (XPO1) with efficacy in various solid and hematologic tumors. We assessed intratumoral penetration, safety, and efficacy of selinexor monotherapy for recurrent glioblastoma. PATIENTS AND METHODS: Seventy-six adults with Karnofsky Performance Status ≥ 60 were enrolled. Patients undergoing cytoreductive surgery received up to three selinexor doses (twice weekly) preoperatively (Arm A; n = 8 patients). Patients not undergoing surgery received 50 mg/m2 (Arm B, n = 24), or 60 mg (Arm C, n = 14) twice weekly, or 80 mg once weekly (Arm D; n = 30). Primary endpoint was 6-month progression-free survival rate (PFS6). RESULTS: Median selinexor concentrations in resected tumors from patients receiving presurgical selinexor was 105.4 nmol/L (range 39.7-291 nmol/L). In Arms B, C, and D, respectively, the PFS6 was 10% [95% confidence interval (CI), 2.79-35.9], 7.7% (95% CI, 1.17-50.6), and 17% (95% CI, 7.78-38.3). Measurable reduction in tumor size was observed in 19 (28%) and RANO-response rate overall was 8.8% [Arm B, 8.3% (95% CI, 1.0-27.0); C: 7.7% (95% CI, 0.2-36.0); D: 10% (95% CI, 2.1-26.5)], with one complete and two durable partial responses in Arm D. Serious adverse events (AEs) occurred in 26 (34%) patients; 1 (1.3%) was fatal. The most common treatment-related AEs were fatigue (61%), nausea (59%), decreased appetite (43%), and thrombocytopenia (43%), and were manageable by supportive care and dose modification. Molecular studies identified a signature predictive of response (AUC = 0.88). CONCLUSIONS: At 80 mg weekly, single-agent selinexor induced responses and clinically relevant PFS6 with manageable side effects requiring dose reductions. Ongoing trials are evaluating safety and efficacy of selinexor in combination with other therapies for newly diagnosed or recurrent glioblastoma.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Hydrazines/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Triazoles/administration & dosage , Administration, Oral , Adult , Aged , Brain/metabolism , Brain Neoplasms/surgery , Cytoreduction Surgical Procedures , Female , Glioblastoma/surgery , Humans , Hydrazines/adverse effects , Hydrazines/metabolism , Male , Middle Aged , Treatment Outcome , Triazoles/adverse effects , Triazoles/metabolism , Young Adult
13.
Clin Transl Sci ; 15(2): 330-342, 2022 02.
Article in English | MEDLINE | ID: mdl-34558200

ABSTRACT

Milvexian (BMS-986177/JNJ-70033093) is a small molecule, active-site inhibitor of factor XIa (FXIa) being developed to prevent and treat thrombotic events. The safety, tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of milvexian were assessed in a two-part, double-blind, placebo-controlled, sequential single ascending dose (SAD) and multiple ascending dose (MAD) study in healthy adults. Participants in SAD panels (6 panels of 8 participants; n = 48) were randomized (3:1) to receive milvexian (4, 20, 60, 200, 300, or 500 mg) or placebo. The 200- and 500-mg panels investigated the pharmacokinetic impact of a high-fat meal. Participants in MAD panels (7 panels of 8 participants; n = 56) were randomized (3:1) to receive milvexian (once- or twice-daily) or placebo for 14 days. All milvexian dosing regimens were safe and well-tolerated, with only mild treatment-emergent adverse events and no clinically significant bleeding events. In SAD panels, maximum milvexian plasma concentration occurred 3 h postdose in all fasted panels. The terminal half-life (T1/2 ) ranged from 8.3 to 13.8 h. In fasted panels from 20 to 200 mg, absorption was dose-proportional; results at higher doses (300 and 500 mg) were consistent with saturable absorption. Food increased milvexian bioavailability in a dose-dependent fashion. In MAD panels, steady-state milvexian plasma concentration was reached within 3 and 6 dosing days with once- and twice-daily dosing, respectively. Renal excretion was less than 20% in all panels. Prolongation of activated partial thromboplastin time was observed and was directly related to drug exposure. These results suggest that the safety, tolerability, PK, and PD properties of milvexian are suitable for further clinical development.


Subject(s)
Factor XIa/drug effects , Pyrimidines/administration & dosage , Triazoles/administration & dosage , Administration, Oral , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Female , Food-Drug Interactions , Humans , Male , Middle Aged , Partial Thromboplastin Time , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Triazoles/pharmacokinetics , Triazoles/pharmacology
14.
J Med Chem ; 65(3): 1770-1785, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34494428

ABSTRACT

Factor XIa (FXIa) is an enzyme in the coagulation cascade thought to amplify thrombin generation but has a limited role in hemostasis. From preclinical models and human genetics, an inhibitor of FXIa has the potential to be an antithrombotic agent with superior efficacy and safety. Reversible and irreversible inhibitors of FXIa have demonstrated excellent antithrombotic efficacy without increased bleeding time in animal models (Weitz, J. I., Chan, N. C. Arterioscler. Thromb. Vasc. Biol. 2019, 39 (1), 7-12). Herein, we report the discovery of a novel series of macrocyclic FXIa inhibitors containing a pyrazole P2' moiety. Optimization of the series for (pharmacokinetic) PK properties, free fraction, and solubility resulted in the identification of milvexian (BMS-986177/JNJ-70033093, 17, FXIa Ki = 0.11 nM) as a clinical candidate for the prevention and treatment of thromboembolic disorders, suitable for oral administration.


Subject(s)
Carotid Artery Thrombosis , Factor XIa , Fibrinolytic Agents , Pyrimidines , Triazoles , Animals , Mice , Rabbits , Administration, Oral , Carotid Artery Thrombosis/drug therapy , Factor XIa/antagonists & inhibitors , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/chemical synthesis , Fibrinolytic Agents/pharmacokinetics , Fibrinolytic Agents/therapeutic use , Macaca fascicularis , Molecular Structure , Pyrazoles/administration & dosage , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Rats, Sprague-Dawley , Structure-Activity Relationship , Triazoles/administration & dosage , Triazoles/chemical synthesis , Triazoles/pharmacokinetics , Triazoles/therapeutic use
15.
Behav Brain Res ; 418: 113644, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34757001

ABSTRACT

Epigenetic mechanisms play important roles in the neurobiology of substance use disorder. In particular, bromodomain and extra-terminal domain (BET) proteins, a class of histone acetylation readers, have been found to regulate cocaine conditioned behaviors, but their role in the behavioral response to other drugs of abuse remains unclear. To address this knowledge gap, we examined the effects of the BET inhibitor, JQ1, on nicotine, amphetamine, morphine, and oxycodone conditioned place preference (CPP). Similar to previous cocaine studies, systemic administration of JQ1 caused a dose-dependent reduction in the acquisition of amphetamine and nicotine CPP in male mice. However, in opioid studies, JQ1 did not alter morphine or oxycodone CPP. Investigating the effects of JQ1 on other types of learning and memory, we found that JQ1 did not alter the acquisition of contextual fear conditioning. Together, these results indicate that BET proteins play an important role in the acquisition of psychostimulant-induced CPP but not the acquisition of opioid-induced CPP nor contextual fear conditioning.


Subject(s)
Anesthetics, Local/pharmacology , Azepines/administration & dosage , Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Triazoles/administration & dosage , Amphetamine/pharmacology , Animals , Cocaine/pharmacology , Epigenomics , Learning/drug effects , Male , Memory/drug effects , Mice , Morphine/pharmacology , Nicotine/pharmacology
16.
J Med Chem ; 65(3): 1767-1769, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34962399

ABSTRACT

Over the past two decades, Factor XIa inhibitors have emerged as an exciting new class of antithrombic agents. Identification of orally bioavailable small molecule inhibitors has presented a formidable challenge for medicinal chemists. Herein, those challenges and the problem-solving that resulted in the discovery of milvexian will be presented.


Subject(s)
Factor XIa/antagonists & inhibitors , Fibrinolytic Agents/therapeutic use , Pyrimidines/therapeutic use , Triazoles/therapeutic use , Administration, Oral , Animals , Drug Discovery , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/chemistry , Humans , Molecular Structure , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Triazoles/administration & dosage , Triazoles/chemistry
17.
Article in English | MEDLINE | ID: mdl-34826613

ABSTRACT

Herbicides pose a potential threat to the soil biodiversity and health. Carfentrazone-ethyl (CE), a triazolinones herbicide, is increasingly used in agricultural production. Its non-target toxic effects on soil microorganisms and soil enzymes are reported recently. However, the sublethal toxicity of CE on soil invertebrates like earthworms is not yet known. Therefore, in this work, the sublethal toxic effects of CE (0.05, 0.5, and 5.0 µg/g in soil) on the soil earthworm (Eisenia fetida) were evaluated using a battery of biomarkers including reactive oxygen species (ROS), enzyme (superoxide dismutase-SOD, catalase-CAT, peroxidase-POD, and glutathione S-transferase-GST) activities, malondialdehyde (MDA) contents, histopathological and DNA damage. Results indicated that CE increased ROS contents, enzyme activities, and MDA contents in the short-time (14 d), thus, causing a slight oxidative stress to E. fetida. However, the toxic effects of CE on earthworms gradually disappeared after 14 days. The CE did not cause histopathological and DNA damage in earthworms. Integrated Biological Response index (IBR) indicated that both concentration and exposure time of CE regulated its sublethal toxicity on earthworms. In conclusion, herbicide CE is safe to soil invertebrate earthworms when applied at the recommended doses. Our results contribute to the current understanding of CE effects on soil earthworms, and can be useful in developing soil health strategies under agrochemical use.


Subject(s)
Herbicides/toxicity , Oligochaeta/drug effects , Triazoles/toxicity , Animals , Ecotoxicology , Enzymes/metabolism , Herbicides/administration & dosage , Intestines/drug effects , Intestines/pathology , Malondialdehyde/metabolism , Oligochaeta/physiology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Soil Pollutants/administration & dosage , Soil Pollutants/toxicity , Triazoles/administration & dosage
20.
Br J Cancer ; 126(5): 718-725, 2022 03.
Article in English | MEDLINE | ID: mdl-34802051

ABSTRACT

BACKGROUND: Proteasome inhibitors (PIs), including carfilzomib, potentiate the activity of selinexor, a novel, first-in-class, oral selective inhibitor of nuclear export (SINE) compound, in preclinical models of multiple myeloma (MM). METHODS: The safety, efficacy, maximum-tolerated dose (MTD) and recommended phase 2 dose (RP2D) of selinexor (80 or 100 mg) + carfilzomib (56 or 70 mg/m2) + dexamethasone (40 mg) (XKd) once weekly (QW) was evaluated in patients with relapsed refractory MM (RRMM) not refractory to carfilzomib. RESULTS: Thirty-two patients, median prior therapies 4 (range, 1-8), were enrolled. MM was triple-class refractory in 38% of patients and 53% of patients had high-risk cytogenetics del(17p), t(4;14), t(14;16) and/or gain 1q. Common treatment-related adverse events (all/Grade 3) were thrombocytopenia 72%/47% (G3 and G4), nausea 72%/6%, anaemia 53%/19% and fatigue 53%/9%, all expected and manageable with supportive care and dose modifications. MTD and RP2D were identified as selinexor 80 mg, carfilzomib 56 mg/m2, and dexamethasone 40 mg, all QW. The overall response rate was 78% including 14 (44%) ≥ very good partial responses. Median progression-free survival was 15 months. CONCLUSIONS: Weekly XKd is highly effective and well-tolerated. These data support further investigation of XKd in patients with MM.


Subject(s)
Dexamethasone/administration & dosage , Hydrazines/administration & dosage , Multiple Myeloma/drug therapy , Oligopeptides/administration & dosage , Triazoles/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone/adverse effects , Drug Administration Schedule , Female , Humans , Hydrazines/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Multiple Myeloma/genetics , Oligopeptides/adverse effects , Survival Analysis , Translocation, Genetic , Treatment Outcome , Triazoles/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...